Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cells ; 12(5)2023 02 22.
Article in English | MEDLINE | ID: mdl-36899830

ABSTRACT

Adenosine 5' triphosphate (ATP) is the energy currency of life, which is produced in mitochondria (~90%) and cytosol (less than 10%). Real-time effects of metabolic changes on cellular ATP dynamics remain indeterminate. Here we report the design and validation of a genetically encoded fluorescent ATP indicator that allows for real-time, simultaneous visualization of cytosolic and mitochondrial ATP in cultured cells. This dual-ATP indicator, called smacATPi (simultaneous mitochondrial and cytosolic ATP indicator), combines previously described individual cytosolic and mitochondrial ATP indicators. The use of smacATPi can help answer biological questions regarding ATP contents and dynamics in living cells. As expected, 2-deoxyglucose (2-DG, a glycolytic inhibitor) led to substantially decreased cytosolic ATP, and oligomycin (a complex V inhibitor) markedly decreased mitochondrial ATP in cultured HEK293T cells transfected with smacATPi. With the use of smacATPi, we can also observe that 2-DG treatment modestly attenuates mitochondrial ATP and oligomycin reduces cytosolic ATP, indicating the subsequent changes of compartmental ATP. To evaluate the role of ATP/ADP carrier (AAC) in ATP trafficking, we treated HEK293T cells with an AAC inhibitor, Atractyloside (ATR). ATR treatment attenuated cytosolic and mitochondrial ATP in normoxia, suggesting AAC inhibition reduces ADP import from the cytosol to mitochondria and ATP export from mitochondria to cytosol. In HEK293T cells subjected to hypoxia, ATR treatment increased mitochondrial ATP along with decreased cytosolic ATP, implicating that ACC inhibition during hypoxia sustains mitochondrial ATP but may not inhibit the reversed ATP import from the cytosol. Furthermore, both mitochondrial and cytosolic signals decrease when ATR is given in conjunction with 2-DG in hypoxia. Thus, real-time visualization of spatiotemporal ATP dynamics using smacATPi provides novel insights into how cytosolic and mitochondrial ATP signals respond to metabolic changes, providing a better understanding of cellular metabolism in health and disease.


Subject(s)
Adenosine Triphosphate , Stress, Physiological , Humans , Cytosol/metabolism , HEK293 Cells , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Atractyloside/metabolism , Oligomycins
2.
Cardiol Rev ; 31(4): 215-218, 2023.
Article in English | MEDLINE | ID: mdl-36730923

ABSTRACT

Reversal of cardiogenic shock depends on its early recognition and prompt initiation of therapy. Recognition of the clinical and hemodynamic deterioration that precedes cardiogenic shock is a crucial step in its early detection. Treatment of pre-cardiogenic shock is chiefly pharmacologic with intravenous administration of pressor, inotropic, and loop diuretic agents. Failure to reverse the preshock state with pharmacotherapy entails progression to cardiogenic shock and the need for prompt mechanical circulatory support with membrane oxygenation and possibly left ventricular decompression.


Subject(s)
Heart-Assist Devices , Shock, Cardiogenic , Humans , Shock, Cardiogenic/diagnosis , Vasoconstrictor Agents/therapeutic use , Hemodynamics , Diuretics/therapeutic use
3.
Am J Med ; 136(1): 27-32, 2023 01.
Article in English | MEDLINE | ID: mdl-36252709

ABSTRACT

Sepsis is an increasing cause of decompensation in patients with chronic heart failure with reduced or preserved ejection fraction. Sepsis and decompensated heart failure results in a mixed septic-cardiogenic shock that poses several therapeutic dilemmas: Rapid fluid resuscitation is the cornerstone of sepsis management, while loop diuretics are the main stay of decompensated heart failure treatment. Whether inotropic therapy with dobutamine or inodilators improves microvascular alterations remains unsettled in sepsis. When to resume loop diuretic therapy in patients with sepsis and decompensated heart failure is unclear. In the absence of relevant guidelines, we review vasopressor therapy, the timing and volume of fluid resuscitation, and the need for inotropic therapy in patients who, with sepsis and decompensated heart failure, present with a mixed septic-cardiogenic shock.


Subject(s)
Heart Failure , Shock, Cardiogenic , Humans , Heart Failure/complications , Heart Failure/therapy
4.
Cells ; 11(9)2022 05 06.
Article in English | MEDLINE | ID: mdl-35563877

ABSTRACT

Previous work showed a role of BNIP3 in myocardial remodeling and progression to HFrEF. We utilized a multiomics approach to unravel BNIP3-related molecular mechanisms in the pathogenesis of HFrEF. BNIP3 knockdown in HFrEF improved glycolysis, pyruvate metabolism, branched-chain amino acid catabolism, and oxidative phosphorylation, and restored endoplasmic reticulum (ER)-mitochondrial (mt) calcium and ion homeostasis. These effects of BNIP3 on cardiac metabolism were related to its interaction and downregulation, and/or phosphorylation, of specific mt-proteins involved in the aforementioned metabolic pathways, including the MICOS and SLC25A families of carrier proteins. BNIP3 affected ER-mt-calcium and ion homeostasis via its interaction-induced VDAC1 dimerization and modulation of VDAC1 phosphorylation at Ser104 and Ser241, and the downregulation of LETM1. At the ER level, BNIP3 interacted with the enzyme SERCA2a and the PKA signaling complex, leading to the downregulation of SERCA2a and PKA-mediated Ser16 phospholamban phosphorylation. Additionally, BNIP3 attenuated AMPK and PRKCE activity by modulating AMPK phosphorylation at Ser485/491 and Ser377 residues, and PRKCE phosphorylation at Thr521 and Thr710 residues. BNIP3 also interacted with sarcomeric, cytoskeletal, and cellular transcription and translation proteins, and affected their expression and/or phosphorylation. In conclusion, BNIP3 modulates multiple pathobiological processes and constitutes an attractive therapeutic target in HFrEF.


Subject(s)
Heart Failure , Ventricular Dysfunction, Left , AMP-Activated Protein Kinases/metabolism , Calcium/metabolism , Heart Failure/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Oxidative Phosphorylation , Proto-Oncogene Proteins/metabolism , Stroke Volume
5.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638917

ABSTRACT

The heart is an organ with high-energy demands in which the mitochondria are most abundant. They are considered the powerhouse of the cell and occupy a central role in cellular metabolism. The intermyofibrillar mitochondria constitute the majority of the three-mitochondrial subpopulations in the heart. They are also considered to be the most important in terms of their ability to participate in calcium and cellular signaling, which are critical for the regulation of mitochondrial function and adenosine triphosphate (ATP) production. This is because they are located in very close proximity with the endoplasmic reticulum (ER), and for the presence of tethering complexes enabling interorganelle crosstalk via calcium signaling. Calcium is an important second messenger that regulates mitochondrial function. It promotes ATP production and cellular survival under physiological changes in cardiac energetic demand. This is accomplished in concert with signaling pathways that regulate both calcium cycling and mitochondrial function. Perturbations in mitochondrial homeostasis and metabolic remodeling occupy a central role in the pathogenesis of heart failure. In this review we will discuss perturbations in ER-mitochondrial crosstalk and touch on important signaling pathways and molecular mechanisms involved in the dysregulation of calcium homeostasis and mitochondrial function in heart failure.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Energy Metabolism/physiology , Heart Failure/metabolism , Adenosine Triphosphate/metabolism , Animals , Endoplasmic Reticulum/metabolism , Heart Failure/pathology , Homeostasis/physiology , Humans , Mitochondria, Heart/metabolism
6.
Int J Mol Sci ; 23(1)2021 Dec 26.
Article in English | MEDLINE | ID: mdl-35008662

ABSTRACT

Metabolic remodeling plays an important role in the pathophysiology of heart failure (HF). We sought to characterize metabolic remodeling and implicated signaling pathways in two rat models of early systolic dysfunction (MOD), and overt systolic HF (SHF). Tandem mass tag-labeled shotgun proteomics, phospho-(p)-proteomics, and non-targeted metabolomics analyses were performed in left ventricular myocardium tissue from Sham, MOD, and SHF using liquid chromatography-mass spectrometry, n = 3 biological samples per group. Mitochondrial proteins were predominantly down-regulated in MOD (125) and SHF (328) vs. Sham. Of these, 82% (103/125) and 66% (218/328) were involved in metabolism and respiration. Oxidative phosphorylation, mitochondrial fatty acid ß-oxidation, Krebs cycle, branched-chain amino acids, and amino acid (glutamine and tryptophan) degradation were highly enriched metabolic pathways that decreased in SHF > MOD. Glycogen and glucose degradation increased predominantly in MOD, whereas glycolysis and pyruvate metabolism decreased predominantly in SHF. PKA signaling at the endoplasmic reticulum-mt interface was attenuated in MOD, whereas overall PKA and AMPK cellular signaling were attenuated in SHF vs. Sham. In conclusion, metabolic remodeling plays an important role in myocardial remodeling. PKA and AMPK signaling crosstalk governs metabolic remodeling in progression to SHF.


Subject(s)
Heart Failure, Systolic/metabolism , Metabolic Networks and Pathways , Metabolomics , Adenylate Kinase/metabolism , Animals , Chromatography, Liquid , Citric Acid Cycle , Cyclic AMP-Dependent Protein Kinases/metabolism , Glycolysis , Mass Spectrometry , Mitochondria/metabolism , Oxidative Phosphorylation , Rats , Signal Transduction
7.
J Clin Med ; 9(11)2020 Nov 06.
Article in English | MEDLINE | ID: mdl-33172082

ABSTRACT

The mitochondria are mostly abundant in the heart, a beating organ of high- energy demands. Their function extends beyond being a power plant of the cell including redox balance, ion homeostasis and metabolism. They are dynamic organelles that are tethered to neighboring structures, especially the endoplasmic reticulum. Together, they constitute a functional unit implicated in complex physiological and pathophysiological processes. Their topology in the cell, the cardiac myocyte in particular, places them at the hub of signaling and calcium homeostasis, making them master regulators of cell survival or cell death. Perturbations in mitochondrial function play a central role in the pathophysiology of myocardial remodeling and progression of heart failure. In this minireview, we summarize important pathophysiological mechanisms, pertaining to mitochondrial morphology, dynamics and function, which take place in compensated hypertrophy and in progression to overt systolic heart failure. Published work in the last few years has expanded our understanding of these important mechanisms; a key prerequisite to identifying therapeutic strategies targeting mitochondrial dysfunction in heart failure.

8.
J Vis Exp ; (158)2020 04 30.
Article in English | MEDLINE | ID: mdl-32420983

ABSTRACT

In response to an injury, such as myocardial infarction, prolonged hypertension or a cardiotoxic agent, the heart initially adapts through the activation of signal transduction pathways, to counteract, in the short-term, for the cardiac myocyte loss and or the increase in wall stress. However, prolonged activation of these pathways becomes detrimental leading to the initiation and propagation of cardiac remodeling leading to changes in left ventricular geometry and increases in left ventricular volumes; a phenotype seen in patients with systolic heart failure (HF). Here, we describe the creation of a rat model of pressure overload induced moderate remodeling and early systolic dysfunction (MOD) by ascending aortic banding (AAB) via a vascular clip with an internal area of 2 mm2. The surgery is performed in 200 g Sprague-Dawley rats. The MOD HF phenotype develops at 8-12 weeks after AAB and is characterized noninvasively by means of echocardiography. Previous work suggests the activation of signal transduction pathways and altered gene expression and post-translational modification of proteins in the MOD HF phenotype that mimic those seen in human systolic HF; therefore, making the MOD HF phenotype a suitable model for translational research to identify and test potential therapeutic anti-remodeling targets in HF. The advantages of the MOD HF phenotype compared to the overt systolic HF phenotype is that it allows for the identification of molecular targets involved in the early remodeling process and the early application of therapeutic interventions. The limitation of the MOD HF phenotype is that it may not mimic the spectrum of diseases leading to systolic HF in human. Moreover, it is a challenging phenotype to create, as the AAB surgery is associated with high mortality and failure rates with only 20% of operated rats developing the desired HF phenotype.


Subject(s)
Disease Models, Animal , Heart Failure, Systolic/physiopathology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Stroke Volume , Ventricular Remodeling/physiology , Animals , Blood Pressure , Echocardiography , Rats , Rats, Sprague-Dawley
9.
Medicina (Kaunas) ; 55(6)2019 Jun 03.
Article in English | MEDLINE | ID: mdl-31163678

ABSTRACT

Background and objectives: Derangements in mitochondrial integrity and function constitute an important pathophysiological feature in the pathogenesis of heart failure (HF) and play an important role in myocardial remodeling and systolic dysfunction. In systolic HF, we and others have shown an imbalance in mitochondrial dynamics toward mitochondrial fission and fragmentation with evidence of mitophagy, mitochondrial vacuolar degeneration, and impairment in mitochondrial oxidative capacity. The morphological stages of mitochondrial vacuolar degeneration have not been defined. We sought to elucidate the progressive stages of mitochondrial vacuolar degeneration, which would serve as a measure to define, morphologically, the severity of mitochondrial damage. Materials and Methods: Transmission electron microscopy was used to study mitochondrial morphology and pathology in phenylephrine-stressed cardiac myocytes in vitro and in left ventricular myocardium from a rat model of pressure overload induced systolic dysfunction and from patients with systolic HF. Results: In phenylephrine-stressed cardiomyocytes for two hours, alterations in mitochondrial cristae morphology (Stage A) and loss and dissolution of mitochondrial cristae in one (Stage B) or multiple (early Stage B→C) mitochondrion area(s) were evident in the earliest stages of mitochondrial vacuolar degeneration. Mitochondrial swelling and progressive dissolution of mitochondrial cristae (advanced Stage B→C), followed by complete loss and dissolution of mitochondrial cristae and permeabilization and destruction of inner mitochondrial membrane (Stage C) then outer mitochondrial membrane rupture (Stage D) constituted advanced stages of mitochondrial vacuolar degeneration. Similar morphological changes in mitochondrial vacuolar degeneration were seen in vivo in animal models and in patients with systolic HF; where about 60-70% of the mitochondria are mainly observed in stages B→C and fewer in stages C and D. Conclusion: Mitochondrial vacuolar degeneration is a prominent mitochondrial morphological feature seen in HF. Defining the progressive stages of mitochondrial vacuolar degeneration would serve as a measure to assess morphologically the severity of mitochondrial damage.


Subject(s)
Heart Failure/complications , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Phenylephrine/adverse effects , Analysis of Variance , Animals , Disease Models, Animal , Heart Failure/physiopathology , Oxidative Stress/drug effects , Phenylephrine/therapeutic use , Rats , Rats, Sprague-Dawley
10.
Circ Heart Fail ; 12(2): e005131, 2019 02.
Article in English | MEDLINE | ID: mdl-30744415

ABSTRACT

BACKGROUND: The FOXO3a (forkhead box O3a)-BNIP3 (B-cell lymphoma 2/adenovirus E1B 19kDa interacting protein 3) pathway modulates mitochondrial dynamics and function and contributes to myocardial remodeling in rodent models of heart failure. We sought to investigate the expression of this pathway along with the expression of mitochondrial biogenesis (PGC-1α [peroxisome proliferator-activated receptor-γ coactivator-1α]), dynamics (DRP-1 [dynamin-related protein 1], OPA-1 [optic atrophy 1], and MFN 2 [mitofusin 2]), and oxidative phosphorylation (citrate synthase and electron transport chain complexes) markers and COX IV (cytochrome C oxidase) activity in myocardium from patients with valvular or ischemic heart disease and heart failure with preserved ejection fraction (HFpEF) or heart failure with reduced ejection fraction (HFrEF). METHODS AND RESULTS: Subepicardial left ventricular biopsies (10×1×1 mm3) were obtained at aortic valve replacement (HFpEFAVR, n=5; and HFrEFAVR, n=4), coronary artery bypass grafting (HFpEFCABG, n=5; and HFrEFCABG, n=5), or left ventricular assist device implantation (HFrEFLVAD, n=4). Subepicardial biopsies from patients with normal left ventricular function (n=2) and from donor hearts (n=3) served as controls (normal). Relative to normal, mitochondrial fragmentation and cristae destruction were evident, and mitochondrial area was decreased in HFpEF; 1.00±0.09 versus 0.71±0.08; P=0.016. These mitochondrial morphological changes were more pronounced in HFrEF (0.54±0.06); P=0.002 HFpEF versus HFrEF. BNIP3 (monomer+dimer) expression was increased in HFpEF (3.99±2.44) and in HFrEF (5.19±1.70) relative to normal; P=0.004 and P<0.001, respectively. However, BNIP3 monomer was increased in HFrEF (4.32±1.43) compared with normal (0.99±0.06) and HFpEF (1.97±0.90); P=0.001 and 0.004, respectively. The HFrEF group uniquely showed increase in DRP-1 expression (1.94±0.38) and decreases in PGC-1α expression (0.61±0.07) and COX IV activity (0.70±0.10) relative to normal; P=0.013, P<0.001, and P<0.001, respectively, with no significant change in electron transport chain complexes expression. CONCLUSIONS: These findings in human myocardium confirm studies in rodents where contractile dysfunction is associated with activation of the FOXO3a-BNIP3 pathway and altered mitochondrial dynamics, biogenesis, and function.


Subject(s)
Forkhead Box Protein O3/metabolism , Heart Failure/metabolism , Membrane Proteins/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Dynamics , Myocardial Ischemia/metabolism , Proto-Oncogene Proteins/metabolism , Stroke Volume , Ventricular Function, Left , Adult , Aged , Aged, 80 and over , Case-Control Studies , Energy Metabolism , Female , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Male , Middle Aged , Mitochondria, Heart/pathology , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Signal Transduction
11.
Methods Mol Biol ; 1816: 195-206, 2018.
Article in English | MEDLINE | ID: mdl-29987821

ABSTRACT

Despite the use of inbred animals, phenotypic variability is usually encountered in rats subjected to pressure overload. This chapter describes techniques for creating a rat model of pressure overload by ascending aortic banding procedure and noninvasive characterization of the variable phenotypes by means of echocardiography. We address the variable phenotypes encountered in this model with moderate versus severe ascending aortic banding. We also describe some of the echocardiographic and hemodynamic parameters and the degree of interstitial fibrosis and extracellular matrix remodeling encountered in each of the different phenotypes.


Subject(s)
Aorta/pathology , Heart Failure/pathology , Heart Ventricles/pathology , Ventricular Remodeling , Animals , Aorta/physiopathology , Blood Pressure , Disease Models, Animal , Disease Progression , Echocardiography , Heart Failure/etiology , Heart Failure/physiopathology , Heart Ventricles/physiopathology , Male , Rats , Rats, Sprague-Dawley
13.
Annu Rev Med ; 69: 65-79, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29414252

ABSTRACT

Heart failure (HF) is a clinical syndrome of diverse etiologies and can be associated with preserved, reduced, or mid-range ejection fraction (EF). In the community, heart failure with preserved ejection fraction (HFpEF) is emerging as the most common form of HF. There remains considerable uncertainty regarding its pathogenesis, diagnosis, and optimal therapeutic approach. Hypotheses have been advanced to explain the underlying pathophysiology responsible for HFpEF, but to date, no specific therapy based on these hypotheses has been proven to improve outcomes in HFpEF. We provide a clinically focused review of the epidemiology, clinical presentation, diagnostic approach, pathophysiology, and treatment of HFpEF.


Subject(s)
Heart Failure/physiopathology , Stroke Volume , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Cardiovascular Agents/therapeutic use , Dyspnea/physiopathology , Exercise Tolerance/physiology , Heart Failure/diagnosis , Heart Failure/drug therapy , Heart Failure/epidemiology , Hospitalization , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypertension/physiopathology , Incidence , Inflammation , Ivabradine/therapeutic use , Mineralocorticoid Receptor Antagonists/therapeutic use , Mortality , Nitrates/therapeutic use , Phosphodiesterase 4 Inhibitors/therapeutic use , Risk Factors
14.
J Am Heart Assoc ; 6(6)2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28619984

ABSTRACT

BACKGROUND: Following pressure overload, compensatory concentric left ventricular remodeling (CR) variably transitions to eccentric remodeling (ER) and systolic dysfunction. Mechanisms responsible for this transition are incompletely understood. Here we leverage phenotypic variability in pressure overload-induced cardiac remodeling to test the hypothesis that altered mitochondrial homeostasis and calcium handling occur early in the transition from CR to ER, before overt systolic dysfunction. METHODS AND RESULTS: Sprague Dawley rats were subjected to ascending aortic banding, (n=68) or sham procedure (n=5). At 3 weeks post-ascending aortic banding, all rats showed CR (left ventricular volumes < sham). At 8 weeks post-ascending aortic banding, ejection fraction was increased or preserved but 3 geometric phenotypes were evident despite similar pressure overload severity: persistent CR, mild ER, and moderate ER with left ventricular volumes lower than, similar to, and higher than sham, respectively. Relative to sham, CR and mild ER phenotypes displayed increased phospholamban, S16 phosphorylation, reduced sodium-calcium exchanger expression, and increased mitochondrial biogenesis/content and normal oxidative capacity, whereas moderate ER phenotype displayed decreased p-phospholamban, S16, increased sodium-calcium exchanger expression, similar degree of mitochondrial biogenesis/content, and impaired oxidative capacity with unique activation of mitochondrial autophagy and apoptosis markers (BNIP3 and Bax/Bcl-2). CONCLUSIONS: After pressure overload, mitochondrial biogenesis and function and calcium handling are enhanced in compensatory CR. The transition to mild ER is associated with decrease in mitochondrial biogenesis and content; however, the progression to moderate ER is associated with enhanced mitochondrial autophagy/apoptosis and impaired mitochondrial function and calcium handling, which precede the onset of overt systolic dysfunction.


Subject(s)
Heart Failure/metabolism , Hypertrophy, Left Ventricular/metabolism , Mitochondria, Heart/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Aorta/physiopathology , Aorta/surgery , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Arterial Pressure , Autophagy , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Heart Failure/etiology , Heart Failure/pathology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Ligation , Mitochondria, Heart/pathology , Organelle Biogenesis , Phosphorylation , Rats, Sprague-Dawley , Ribosomal Proteins/metabolism , Sodium-Calcium Exchanger/metabolism , Time Factors , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
15.
Am J Physiol Heart Circ Physiol ; 311(6): H1540-H1559, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27694219

ABSTRACT

The forkhead box O3a (FOXO3a) transcription factor has been shown to regulate glucose metabolism, muscle atrophy, and cell death in postmitotic cells. Its role in regulation of mitochondrial and myocardial function is not well studied. Based on previous work, we hypothesized that FOXO3a, through BCL2/adenovirus E1B 19-kDa protein-interacting protein 3 (BNIP3), modulates mitochondrial morphology and function in heart failure (HF). We modulated the FOXO3a-BNIP3 pathway in normal and phenylephrine (PE)-stressed adult cardiomyocytes (ACM) in vitro and developed a cardiotropic adeno-associated virus serotype 9 encoding dominant-negative FOXO3a (AAV9.dn-FX3a) for gene delivery in a rat model of HF with preserved ejection fraction (HFpEF). We found that FOXO3a upregulates BNIP3 expression in normal and PE-stressed ACM, with subsequent increases in mitochondrial Ca2+, leading to decreased mitochondrial membrane potential, mitochondrial fragmentation, and apoptosis. Whereas dn-FX3a attenuated the increase in BNIP3 expression and its consequences in PE-stressed ACM, AAV9.dn-FX3a delivery in an experimental model of HFpEF decreased BNIP3 expression, reversed adverse left ventricular remodeling, and improved left ventricular systolic and, particularly, diastolic function, with improvements in mitochondrial structure and function. Moreover, AAV9.dn-FX3a restored phospholamban phosphorylation at S16 and enhanced dynamin-related protein 1 phosphorylation at S637. Furthermore, FOXO3a upregulates maladaptive genes involved in mitochondrial apoptosis, autophagy, and cardiac atrophy. We conclude that FOXO3a activation in cardiac stress is maladaptive, in that it modulates Ca2+ cycling, Ca2+ homeostasis, and mitochondrial dynamics and function. Our results suggest an important role of FOXO3a in HF, making it an attractive potential therapeutic target.


Subject(s)
Calcium/metabolism , Forkhead Box Protein O3/genetics , Heart Failure/metabolism , Membrane Proteins/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Blotting, Western , Calcium-Binding Proteins/metabolism , Cell Survival , Citrate (si)-Synthase/metabolism , Disease Models, Animal , Dynamins/metabolism , Echocardiography , Electron Transport Complex IV/metabolism , Endoplasmic Reticulum/metabolism , Fluorescent Antibody Technique , Forkhead Box Protein O3/metabolism , Heart Failure/physiopathology , In Vitro Techniques , Male , Membrane Potential, Mitochondrial , Microscopy, Electron, Transmission , Mitochondria, Heart/drug effects , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Phenylephrine/pharmacology , Phosphorylation , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Stress, Physiological , Stroke Volume , Sympathomimetics/pharmacology , Ventricular Function, Left/genetics , Ventricular Remodeling
16.
Physiol Rep ; 3(7)2015 Jul.
Article in English | MEDLINE | ID: mdl-26152691

ABSTRACT

Autophagy, macroautophagy and chaperone-mediated autophagy (CMA), are upregulated in pressure overload (PO) hypertrophy. In this study, we targeted this process at its induction using 3 methyladenine and at the lysosomal level using chloroquine and evaluated the effects of these modulations on cardiac function and myocyte ultrastructure. Sprague-Dawley rats weighing 200 g were subjected to ascending aortic banding. After 1 week of PO, animals were randomized to receive 3 methyladenine versus chloroquine, intraperitoneally, for 2 weeks at a dose of 40 and 50 mg/kg/day, respectively. Saline injection was used as control. Chloroquine treatment, in PO, resulted in regression in cardiac hypertrophy but with significant impairments in cardiac relaxation and contractility. Ultrastructurally, chloroquine accentuated mitochondrial fragmentation and cristae destruction with a plethora of autophagosomes containing collapsed mitochondria and lysosomal lamellar bodies. In contrast, 3 methyladenine improved cardiac function and attenuated mitochondrial fragmentation and autophagososme formation. Markers of macroautophagy and CMA were significantly decreased in the chloroquine group; whereas 3 methyladenine treatment significantly attenuated macroautophagy with a compensatory increase in CMA. Furthermore, chloroquine accentuated PO induced oxidative stress through the further decrease in the expression of manganese superoxide dismutase; whereas, 3 MA had a completely opposite effect. Taken together, these data suggest that high-dose chloroquine, in addition to its effect on the autophagy-lysosome pathway, significantly impairs mitochondrial antioxidant buffering capacity and accentuates oxidative stress and mitochondrial dysfunction in PO hypertrophy; highlighting, the cautious administration of this drug in high oxidative stress conditions, such as pathological hypertrophy or heart failure.

17.
Curr Heart Fail Rep ; 11(4): 354-65, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224319

ABSTRACT

The prevalence of heart failure (HF) and its subtype, HF with preserved ejection fraction (HFpEF), is on the rise due to aging of the population. HFpEF is convergence of several pathophysiological processes, which are not yet clearly identified. HFpEF is usually seen in association with systemic diseases, such as diabetes, hypertension, atrial fibrillation, sleep apnea, renal and pulmonary disease. The proportion of HF patients with HFpEF varies by patient demographics, study settings (cohort vs. clinical trial, outpatient clinics vs. hospitalised patients) and cut points used to define preserved function. There is an expanding body of literature about prevalence and prognostic significance of both cardiovascular and non-cardiovascular comorbidities in HFpEF patients. Current therapeutic approaches are targeted towards alleviating the symptoms, treating the associated comorbid conditions, and reducing recurrent hospital admissions. There is lack of evidence-based therapies that show a reduction in the mortality amongst HFpEF patients; however, an improvement in exercise tolerance and quality of life is seen with few interventions. In this review, we highlight the epidemiology and current treatment options for HFpEF.


Subject(s)
Heart Failure/epidemiology , Heart Failure/physiopathology , Stroke Volume/physiology , Comorbidity , Demography , Edema/physiopathology , Edema/prevention & control , Heart Failure/diagnosis , Heart Failure/therapy , Heart Rate/physiology , Hospitalization/statistics & numerical data , Humans , Hypertension/physiopathology , Hypertension/prevention & control , Incidence , Myocardial Ischemia/physiopathology , Myocardial Ischemia/prevention & control , Prevalence , Risk Factors
18.
Gene Ther ; 21(4): 379-386, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24572786

ABSTRACT

Adeno-associated virus (AAV)-based vectors are promising vehicles for therapeutic gene delivery, including for the treatment for heart failure. It has been demonstrated for each of the AAV serotypes 1 through 8 that inhibition of the proteasome results in increased transduction efficiencies. For AAV9, however, the effect of proteasome inhibitors on in vivo transduction has until now not been evaluated. Here we demonstrate, in a well-established rodent heart failure model, that concurrent treatment with the proteasome inhibitor bortezomib does not enhance the efficacy of AAV9.SERCA2a to improve cardiac function as examined by echocardiography and pressure volume analysis. Western blot analysis of SERCA2a protein and reverse transcription-PCR of SERCA2a mRNA demonstrated that bortezomib had no effect on either endogenous rat SERCA2a levels nor on expression levels of human SERCA2a delivered by AAV9.SERCA2a. Similarly, the number of AAV9 genomes in heart samples was unaffected by bortezomib treatment. Interestingly, whereas transduction of HeLa cells and neonatal rat cardiomyocytes by AAV9 was stimulated by bortezomib, transduction of adult rat cardiomyocytes was inhibited. These results indicate an organ/cell-type-specific effect of proteasome inhibition on AAV9 transduction. A future detailed analysis of the underlying molecular mechanisms promises to facilitate the development of improved AAV vectors.


Subject(s)
Boronic Acids/administration & dosage , Genetic Therapy , Heart Failure/therapy , Myocytes, Cardiac/metabolism , Pyrazines/administration & dosage , Animals , Bortezomib , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Heart Failure/genetics , Humans , Myocardium/pathology , Myocytes, Cardiac/drug effects , Rats , Sarcoplasmic Reticulum Calcium-Transporting ATPases/biosynthesis
19.
J Am Heart Assoc ; 2(2): e000078, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23612897

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) play a key role in the development of heart failure, and recent studies have shown that the muscle-specific miR-1 is a key regulator of cardiac hypertrophy. We tested the hypothesis that chronic restoration of miR-1 gene expression in vivo will regress hypertrophy and protect against adverse cardiac remodeling induced by pressure overload. METHODS AND RESULTS: Cardiac hypertrophy was induced by left ventricular pressure overload in male Sprague-Dawley rats subjected to ascending aortic stenosis. When the hypertrophy was established at 2 weeks after surgery, the animals were randomized to receive either an adeno-associated virus expressing miR-1 (AAV9.miR-1) or green fluorescent protein (GFP) as control (AAV9.GFP) via a single-bolus tail-vein injection. Administration of miR-1 regressed cardiac hypertrophy (left ventricular posterior wall thickness,; 2.32±0.08 versus 2.75±0.07 mm, P<0.001) and (left ventricular septum wall thickness, 2.23±0.06 versus 2.54±0.10 mm, P<0.05) and halted the disease progression compared with control-treated animals, as assessed by echocardiography (fractional shortening, 37.60±5.01% versus 70.68±2.93%, P<0.05) and hemodynamic analyses (end-systolic pressure volume relationship/effective arterial elastance, 1.87±0.46 versus 0.96±0.38, P<0.05) after 7 weeks of treatment. Additionally, miR-1 replacement therapy lead to a marked reduction of myocardial fibrosis, an improvement in calcium handling, inhibition of apoptosis, and inactivation of the mitogen-activated protein kinase signaling pathways, suggesting a favorable effect on preventing the maladaptive ventricular remodeling. We also identified and validated a novel bona fide target of miR-1, Fibullin-2 (Fbln2), a secreted protein implicated in extracellular matrix remodeling. CONCLUSIONS: Taken together, our findings suggest that restoration of miR-1 gene expression is a potential novel therapeutic strategy to reverse pressure-induced cardiac hypertrophy and prevent maladaptive cardiac remodeling.


Subject(s)
Calcium-Binding Proteins/physiology , Extracellular Matrix Proteins/physiology , Hypertrophy, Left Ventricular/therapy , MicroRNAs/therapeutic use , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Ventricular Remodeling , Animals , Apoptosis , Calcium-Binding Proteins/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/therapy , Extracellular Matrix Proteins/metabolism , Fibrosis , Gene Expression Profiling , Gene Transfer Techniques , Genetic Therapy , Humans , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Male , MicroRNAs/genetics , Mitogen-Activated Protein Kinase Kinases , Myocardium/pathology , Myocytes, Cardiac/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction
20.
Circ Heart Fail ; 6(3): 572-83, 2013 May.
Article in English | MEDLINE | ID: mdl-23508759

ABSTRACT

BACKGROUND: We have shown that BNIP3 expression is significantly increased in heart failure (HF). In this study, we tested the effects of BNIP3 manipulation in HF. METHODS AND RESULTS: In a rat model of pressure overload HF, BNIP3 knockdown significantly decreased left ventricular (LV) volumes with significant improvement in LV diastolic and systolic function. There were significant decreases in myocardial apoptosis and LV interstitial fibrosis. Ultrastructurally, BNIP3 knockdown attenuated mitochondrial fragmentation and restored mitochondrial morphology and integrity. On the molecular level, there were significant decreases in endoplasmic reticulum (ER) stress and mitochondrial apoptotic markers. One of the mechanisms by which BNIP3 mediates mitochondrial dysfunction is via the oligomerization of the voltage-dependent anion channels causing a shift of calcium from the ER to mitochondrial compartments, leading to the decrease in ER calcium content, mitochondrial damage, apoptosis, and LV interstitial fibrosis, and hence contributes to both systolic and diastolic myocardial dysfunction, respectively. In systolic HF, the downregulation of SERCA2a (sarcoplasmic-endoplasmic reticulum calcium ATPase), along with an increased BNIP3 expression, further worsen myocardial diastolic and systolic function and contribute to the major remodeling seen in systolic HF as compared with diastolic HF with normal SERCA2a expression. CONCLUSIONS: The increase in BNIP3 expression contributes mainly to myocardial diastolic dysfunction through mitochondrial apoptosis, LV interstitial fibrosis, and to some extent to myocardial systolic dysfunction attributable to the shift of calcium from the ER to the mitochondria and to the decrease in ER calcium content. However, SERCA2a downregulation remains a prerequisite for the major LV remodeling seen in systolic HF.


Subject(s)
Calcium/physiology , Heart Failure, Diastolic/physiopathology , Heart Failure, Systolic/physiopathology , Membrane Proteins/physiology , Mitochondria, Heart/metabolism , Myocytes, Cardiac/physiology , Proto-Oncogene Proteins/physiology , Sarcoplasmic Reticulum/physiology , Adenoviridae/genetics , Animals , Apoptosis/physiology , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Homeostasis , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Voltage-Dependent Anion Channel 1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...